Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • On the Market
  • Published:

Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig–coated artificial antigen-presenting cells

Abstract

Adoptive immunotherapy holds promise as a treatment for cancer and infectious diseases, but its development has been impeded by the lack of reproducible methods for generating therapeutic numbers of antigen-specific CD8+ cytotoxic T lymphocytes (CTLs). As a result, there are only limited reports of expansion of antigen-specific CTLs to the levels required for clinical therapy. To address this issue, artificial antigen-presenting cells (aAPCs) were made by coupling a soluble human leukocyte antigen–immunoglobulin fusion protein (HLA-Ig) and CD28-specific antibody to beads. HLA-Ig–based aAPCs were used to induce and expand CTLs specific for cytomegalovirus (CMV) or melanoma. aAPC-induced cultures showed robust antigen-specific CTL expansion over successive rounds of stimulation, resulting in the generation of clinically relevant antigen-specific CTLs that recognized endogenous antigen–major histocompatibility complex complexes presented on melanoma cells. These studies show the value of HLA-Ig–based aAPCs for reproducible expansion of disease-specific CTLs for clinical approaches to adoptive immunotherapy.

This is a preview of subscription content, access via your institution

Relevant articles

Open Access articles citing this article.

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Schematic of induction and expansion of peptide-specific CTLs by autologous DCs or aAPCs.
Figure 2: Induction and growth potential of Mart-1–specific CD8+ T cells stimulated with aAPCs.
Figure 3: aAPC-induced antigen-specific CTLs recognize endogenous melanoma or CMV pp65 antigen on target cells.
Figure 4: Frequency of antigen-specific CTLs after expansion with CD3-specific antibody beads or aAPCs.

References

  1. Walter, E.A. et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N. Engl. J. Med. 333, 1038–1044 (1995).

    Article  CAS  Google Scholar 

  2. Heslop, H.E. et al. Long-term restoration of immunity against Epstein-Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes. Nat. Med. 2, 551–555 (1996).

    Article  CAS  Google Scholar 

  3. Aebersold, P. et al. Lysis of autologous melanoma cells by tumor-infiltrating lymphocytes: association with clinical response. J. Natl. Cancer Inst. 83, 932–937 (1991).

    Article  CAS  Google Scholar 

  4. Levine, B.L. et al. Large-scale production of CD4+ T cells from HIV-1-infected donors after CD3/CD28 costimulation. J. Hematother. 7, 437–448 (1998).

    Article  CAS  Google Scholar 

  5. Deeths, M.J. & Mescher, M.F. B7-1-dependent co-stimulation results in qualitatively and quantitatively different responses by CD4+ and CD8+ T cells. Eur. J. Immunol. 27, 598–608 (1997).

    Article  CAS  Google Scholar 

  6. Maus, M.V. et al. Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs expressing ligands for the T-cell receptor, CD28 and 4-1BB. Nat. Biotechnol. 20, 143–148 (2002).

    Article  CAS  Google Scholar 

  7. Greten, T.F. et al. Direct visualization of antigen-specific T cells: HTLV-1 Tax11-19-specific CD8+ T cells are activated in peripheral blood and accumulate in cerebrospinal fluid from HAM/TSP patients. Proc. Natl. Acad. Sci. USA 95, 7568–7573 (1998).

    Article  CAS  Google Scholar 

  8. Valmori, D. et al. Optimal activation of tumor-reactive T cells by selected antigenic peptide analogues. Int. Immunol. 11, 1971–1980 (1999).

    Article  CAS  Google Scholar 

  9. Oelke, M. et al. Generation and purification of CD8+ melan-A-specific cytotoxic T lymphocytes for adoptive transfer in tumor immunotherapy. Clin. Cancer Res. 6, 1997–2005 (2000).

    CAS  PubMed  Google Scholar 

  10. von Bergwelt-Baildon, M.S. et al. Human primary and memory cytotoxic T lymphocyte responses are efficiently induced by means of CD40-activated B cells as antigen-presenting cells: potential for clinical application. Blood 99, 3319–3325 (2002).

    Article  CAS  Google Scholar 

  11. Jager, E. et al. Simultaneous humoral and cellular immune response against cancer-testis antigen NY-ESO-1: definition of human histocompatibility leukocyte antigen (HLA)-A2-binding peptide epitopes. J. Exp. Med. 187, 265–270 (1998).

    Article  CAS  Google Scholar 

  12. Lee, S.P. et al. HLA A2.1-restricted cytotoxic T cells recognizing a range of Epstein-Barr virus isolates through a defined epitope in latent membrane protein LMP2. J. Virol. 67, 7428–7435 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Yee, C., Savage, P.A., Lee, P.P., Davis, M.M. & Greenberg, P.D. Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide-MHC tetramers. J. Immunol. 162, 2227–2234 (1999).

    CAS  PubMed  Google Scholar 

  14. Salgaller, M.L. et al. Report of immune monitoring of prostate cancer patients undergoing T-cell therapy using dendritic cells pulsed with HLA-A2-specific peptides from prostate-specific membrane antigen (PSMA). Prostate 35, 144–151 (1998).

    Article  CAS  Google Scholar 

  15. Laux, I. et al. Response differences between human CD4(+) and CD8(+) T-cells during CD28 costimulation: implications for immune cell-based therapies and studies related to the expansion of double-positive T-cells during aging. Clin. Immunol. 96, 187–197 (2000).

    Article  CAS  Google Scholar 

  16. Tham, E.L., Jensen, P.L. & Mescher, M.F. Activation of antigen-specific T cells by artificial cell constructs having immobilized multimeric peptide-class I complexes and recombinant B7-Fc proteins. J. Immunol. Methods 249, 111–119 (2001).

    Article  CAS  Google Scholar 

  17. Latouche, J.B. & Sadelain, M. Induction of human cytotoxic T lymphocytes by artificial antigen-presenting cells. Nat. Biotechnol. 18, 405–409 (2000).

    Article  CAS  Google Scholar 

  18. Oelke, M. et al. Functional characterization of CD8(+) antigen-specific cytotoxic T lymphocytes after enrichment based on cytokine secretion: comparison with the MHC-tetramer technology. Scand. J. Immunol. 52, 544–549 (2000).

    Article  CAS  Google Scholar 

  19. Perez-Diez, A. et al. Generation of CD8+ and CD4+ T-cell response to dendritic cells genetically engineered to express the MART-1/Melan-A gene. Cancer Res. 58, 5305–5309 (1998).

    CAS  PubMed  Google Scholar 

  20. Valmori, D. et al. Vaccination with a Melan-A peptide selects an oligoclonal T cell population with increased functional avidity and tumor reactivity. J. Immunol. 168, 4231–4240 (2002).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Laumer, J. Heymann and S. Vogel for technical assistance and G. Hawkins and J. Zaia for providing the A293 cell lines. Support for this work was provided by National Institutes of Health grants AI-29575 and AI-44129, the Dr. Mildred-Scheel-Stiftung Deutsche Krebshilfe Foundation (M.O.), the Abramson Cancer Research Institute (C.H.J.) and NIH training grant DK07748 (M.V.M.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mathias Oelke.

Ethics declarations

Competing interests

Under licensing agreements between BD-Pharmingen and Johns Hopkins University, J.P.S. is entitled to a share of the royalty received by the University on sales of DimerX products. J.P.S. is also a consultant to BD-Pharmingen. The terms of this agreement are being managed by Johns Hopkins University in accordance with its conflict-of-interest policies.

C.H.J. is an inventor on a patent describing a related technology using anti-CD28 on polystyrene paramagnetic beads (US #6,352,694). Under licensing agreements between the United States Government and Wyeth and Xcyte Therapies, Inc., C.H.J. is entitled to a share of the royalty received by the government for this related bead technology. The terms of this agreement are managed by the Office of Naval Research in accordance with the United States Government conflict-of-interest policies.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Cite this article

Oelke, M., Maus, M., Didiano, D. et al. Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig–coated artificial antigen-presenting cells. Nat Med 9, 619–625 (2003). https://doi.org/10.1038/nm869

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm869

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing